The HSPA12A antibody, FITC conjugated is optimized for:
Immunofluorescence (IF): Detection of HSPA12A in fixed and permeabilized cells or tissue sections.
Flow Cytometry: Quantitative analysis of HSPA12A expression in cell suspensions.
Western Blot (WB): While not explicitly validated for WB by Abbexa, other HSPA12A antibodies (e.g., Abcam’s monoclonal variant) are compatible with WB .
Example Use Case: In studies of hepatocyte stress responses, this antibody could localize HSPA12A expression in liver tissue sections using confocal microscopy .
HSPA12A is a stress-inducible chaperone protein implicated in:
Liver Injury: Attenuates lipopolysaccharide (LPS)-induced hepatocyte pyroptosis via PGC-1α signaling .
Cancer Progression: Modulates CD147 stability to suppress tumor glycolysis and migration .
Cardiac Fibrosis: Acts as a scaffolding protein to regulate p53 ubiquitination and fibroblast activation .
While the antibody itself is not the focus of these studies, its availability enables precise visualization of HSPA12A in experimental models, facilitating mechanistic insights .
The following table highlights key differences among commercially available HSPA12A antibodies:
Catalog Number | Conjugate | Clonality | Applications | Reactivity |
---|---|---|---|---|
Abbexa (FITC) | FITC | Polyclonal | IF/ICC, Flow Cytometry | Human |
Abcam (ab200838) | Unconjugated | Monoclonal | IP, WB, IHC-P, IF | Human, Mouse, Rat |
Antibodies-Online | HRP | Polyclonal | ELISA | Human |
HSPA12A (Heat Shock Protein Family A Member 12A) is a novel member of the HSP70 family with emerging roles in multiple pathological processes. Unlike conventional heat shock proteins, HSPA12A demonstrates unique biological functions including: binding to PCNA to promote its trimerization in hepatocellular carcinoma , interacting with PGC-1α to regulate AOAH expression and prevent hepatocyte pyroptosis , controlling cerebral lactate homeostasis , and regulating HMGB1 lactylation and secretion in liver ischemia/reperfusion injury . This multifunctionality makes HSPA12A a significant research target across cancer biology, liver pathology, and neurological disorders.
FITC-conjugated HSPA12A antibodies contain the fluorescein isothiocyanate fluorophore directly attached to the antibody molecule, allowing direct visualization without secondary antibodies. This provides several methodological advantages:
Single-step detection process in immunofluorescence and flow cytometry
Elimination of cross-reactivity issues that can occur with secondary antibodies
Consistent signal-to-noise ratio across experiments
Compatibility with multicolor immunofluorescence when combined with antibodies conjugated to spectrally distinct fluorophores
Methodologically, researchers should recognize that FITC conjugation may slightly alter antibody binding kinetics compared to unconjugated antibodies, potentially requiring optimization of antibody concentration in experimental protocols.
For rigorous research applications, the following experimental controls are essential:
These controls ensure that observed signals represent genuine HSPA12A detection rather than technical artifacts, particularly important when publishing novel HSPA12A localization or interaction findings.
HSPA12A promotes hepatocellular carcinoma growth through direct binding to proliferating cell nuclear antigen (PCNA) and enhancing its trimerization, which is essential for its function in carcinogenesis . To investigate this interaction:
Co-localization studies: Use FITC-conjugated HSPA12A antibody alongside a spectrally distinct fluorophore-conjugated anti-PCNA antibody to visualize potential co-localization in HCC cells using confocal microscopy.
Proximity ligation assay (PLA): Combine FITC-conjugated HSPA12A antibody with PCNA antibody in PLA to detect and quantify protein interactions at single-molecule resolution.
FRET analysis: Pair FITC-conjugated HSPA12A antibody with a compatible FRET acceptor-conjugated PCNA antibody to measure interaction dynamics in live cells.
Immunoprecipitation verification: Following co-immunoprecipitation using anti-HSPA12A or anti-PCNA antibodies, use FITC-conjugated antibodies for detection in Western blotting, similar to approaches used in the original studies demonstrating this interaction .
These approaches allow researchers to investigate whether experimental manipulations (drug treatments, genetic modifications) affect the HSPA12A-PCNA interaction that drives HCC progression.
Studies show HSPA12A promotes β-catenin expression and nuclear translocation in HCC . To investigate this mechanism:
Subcellular fractionation protocol:
Separate nuclear and cytoplasmic fractions from HCC cells using differential centrifugation
Quantify HSPA12A and β-catenin in each fraction via immunoblotting
Use FITC-conjugated HSPA12A antibody for direct visualization in immunofluorescence studies
Live-cell imaging approach:
Co-transfect cells with tagged β-catenin and modulate HSPA12A expression
Track dynamic changes in β-catenin localization using time-lapse microscopy
Correlate with HSPA12A expression patterns using fixed-cell immunofluorescence
Quantification methods:
Measure nuclear/cytoplasmic β-catenin ratios using digital image analysis
Compare ratios between HSPA12A-overexpressing and knockdown conditions
Establish temporal relationship between HSPA12A expression and β-catenin translocation
This systematic approach can determine whether HSPA12A's effect on β-catenin is direct or involves intermediate signaling molecules.
HSPA12A attenuates lipopolysaccharide-induced liver injury by inhibiting caspase-11-mediated hepatocyte pyroptosis via PGC-1α-dependent AOAH expression . To investigate this protective mechanism:
Dual immunofluorescence protocol:
Co-stain liver sections or cultured hepatocytes with FITC-conjugated HSPA12A antibody and markers of pyroptosis (e.g., anti-GSDMD-N)
Analyze spatial relationships between HSPA12A expression and pyroptotic cells
Quantify correlation between HSPA12A intensity and pyroptosis markers
LPS tracing experiments:
Mechanistic studies:
Visualize HSPA12A-PGC-1α interactions through proximity ligation or co-immunoprecipitation
Track PGC-1α nuclear translocation in relation to HSPA12A expression
Correlate with downstream AOAH expression and activity
This approach helps establish the molecular sequence from HSPA12A expression to hepatoprotection through reduction of pyroptotic cell death.
HSPA12A protects against liver ischemia/reperfusion injury by suppressing glycolysis-derived lactate production, thereby inhibiting HMGB1 lactylation and exosomal secretion . A comprehensive experimental design would include:
In vivo protocol:
Establish liver ischemia/reperfusion injury in hepatocyte-specific HSPA12A overexpression mice (h-Ki) and controls
Collect serum samples at multiple timepoints to measure HMGB1 levels by ELISA
Process liver tissue for immunofluorescence using FITC-conjugated HSPA12A antibody alongside HMGB1 staining
In vitro hypoxia/reoxygenation (H/R) system:
Subject primary hepatocytes with manipulated HSPA12A expression to H/R conditions
Analyze intracellular HMGB1 lactylation by immunoprecipitation followed by anti-Klac immunoblotting
Isolate exosomes from culture medium to quantify secreted HMGB1
Glycolytic analysis:
This multi-level approach allows for comprehensive evaluation of the HSPA12A→glycolysis→lactate→HMGB1 lactylation→secretion pathway in liver protection.
HSPA12A controls cerebral lactate homeostasis to maintain mood stability through inhibiting GSK3β in hippocampal neurons . A systematic experimental approach would include:
Brain region-specific analysis:
Section mouse brain tissue from wildtype and Hspa12a-/- animals
Perform immunofluorescence with FITC-conjugated HSPA12A antibody and neuronal markers
Quantify regional distribution patterns with focus on hippocampus
Primary neuron culture studies:
Isolate hippocampal neurons from mice with different HSPA12A expression
Track intracellular lactate using fluorescent probes while monitoring HSPA12A localization
Induce GSK3β activation and observe effects on lactate production
In vivo recovery experiments:
Quantitative analysis:
Develop co-registration techniques for HSPA12A signal and lactate measurements
Implement computational approaches to correlate spatial patterns
Apply statistical methods to determine significance of observed relationships
This comprehensive approach links molecular mechanisms to physiological outcomes in the context of mood regulation.
HSPA12A unstabilizes CD147 to inhibit lactate export and migration in renal cell carcinoma . To demonstrate this relationship:
Protein stability assays:
Ubiquitination analysis:
Immunoprecipitate CD147 from cells with manipulated HSPA12A expression
Probe for ubiquitin to assess ubiquitination status
Identify interaction with E3 ligase HRD1 through co-immunoprecipitation studies
Lactate export measurements:
Measure extracellular acidification rate using Seahorse technology
Correlate with HSPA12A expression determined by flow cytometry using FITC-conjugated antibodies
Rescue experiments by overexpressing CD147 in HSPA12A-overexpressing cells
Migration assays:
Perform wound healing and Transwell migration assays under different HSPA12A conditions
Visualize HSPA12A localization during migration using FITC-conjugated antibodies
Correlate migration indices with HSPA12A and CD147 expression levels
This systematic approach establishes the complete mechanism from HSPA12A expression to CD147 degradation to functional consequences in RCC.
Epitope masking can occur when protein-protein interactions block antibody access to HSPA12A epitopes, particularly problematic when studying HSPA12A's interactions with PCNA , PGC-1α , or HRD1 . To address this methodological challenge:
Epitope mapping approach:
Test multiple FITC-conjugated HSPA12A antibodies targeting different epitopes
Create a panel of antibodies recognizing N-terminal, central domain, and C-terminal regions
Compare detection efficiency in contexts where specific interactions are known to occur
Mild fixation optimization:
Test gradient of fixation conditions (from 0.1-4% paraformaldehyde)
Compare results with methanol fixation which may preserve different epitopes
Consider antigen retrieval methods to expose masked epitopes
Proximity-based alternatives:
When direct epitope detection fails, implement proximity ligation assays
Use protein tagging approaches (FLAG, HA) on recombinant HSPA12A
Consider split-GFP complementation to visualize interactions without antibodies
This methodical approach ensures detection of HSPA12A even when engaged in complex interactions that might mask conventional antibody binding sites.
Liver and brain tissues often present high autofluorescence in the FITC emission spectrum, creating technical challenges for specific signal detection. Recommended solutions include:
Spectral unmixing approach:
Capture full emission spectra from labeled and unlabeled tissue samples
Use computational algorithms to separate HSPA12A-specific signal from autofluorescence
Apply consistent unmixing parameters across experimental groups
Autofluorescence quenching protocol:
Pretreat tissue sections with Sudan Black B (0.1% in 70% ethanol)
Apply copper sulfate (10mM CuSO₄ in 50mM ammonium acetate buffer)
Optimize treatment duration to maximize quenching while preserving antibody binding
Alternative detection strategies:
Consider antibodies conjugated to fluorophores with longer emission wavelengths
Implement tyramide signal amplification for stronger specific signals
Use quantum dots for superior photostability and brightness
Quantification approach:
Develop ratiometric analysis comparing signal in regions of interest to background
Apply tissue-specific threshold algorithms based on controls
Implement machine learning for automated signal/background discrimination
These techniques ensure reliable detection of HSPA12A even in challenging tissue environments with high background fluorescence.
When investigating HSPA12A's interactions with PCNA , PGC-1α , HRD1 , or HMGB1 , optimized co-detection is essential:
Sequential staining protocol:
Apply primary antibodies sequentially rather than simultaneously
Begin with the weaker signal target, typically the interaction partner
Use FITC-conjugated HSPA12A antibody in the second round of staining
Block between rounds with excess unconjugated antibody from the same species
Signal amplification strategy:
Apply tyramide signal amplification for the weaker interaction partner
Balance signal intensities through exposure optimization
Consider spectral overlap and select compatible fluorophores
Sample preparation optimization:
Test multiple fixation methods to preserve both protein epitopes
Optimize permeabilization to balance access to nuclear/cytoplasmic targets
Validate with individual staining before attempting co-detection
Advanced imaging setup:
Implement sequential scanning on confocal microscopy to minimize bleed-through
Use appropriate negative controls to set detection thresholds
Apply deconvolution algorithms to enhance signal separation
This systematic approach enables reliable co-detection of HSPA12A and its binding partners across experimental conditions.
For rigorous quantification of HSPA12A in models like hepatocellular carcinoma , liver injury , or renal cell carcinoma :
Western blot quantification method:
Normalize HSPA12A signal to multiple loading controls (e.g., β-actin, GAPDH)
Apply densitometry using software that corrects for non-linear response
Present data as fold change relative to appropriate controls
Include technical and biological replicates (n≥3) for statistical validity
Flow cytometry approach:
Establish negative and positive controls to set gating parameters
Report results as mean fluorescence intensity rather than percent positive
Apply compensation when using multiple fluorophores
Consider cells expressing varying HSPA12A levels as internal controls
Image analysis protocol:
Define regions of interest based on biological relevance (e.g., nuclear vs. cytoplasmic)
Apply consistent thresholding algorithms across all experimental conditions
Quantify both signal intensity and pattern distribution
Present data with appropriate statistical tests for significance
These approaches provide robust quantification of HSPA12A expression changes, allowing for reliable interpretation of experimental results across different disease models.